1
|
de Oliveira Costa M, Dame MK. In Vitro Porcine (Explant) Colon Culture. Methods Mol Biol 2024; 2749:91-101. [PMID: 38133777 DOI: 10.1007/978-1-0716-3609-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Models have been extensively used to investigate disease pathogenesis. Animal models are costly and require extensive logistics for animal care, and samples are not always suitable for different analytical techniques or to answer the research question. In vitro cell culture models are generally focused on recreating a specific characteristic of an organ and are limited to a single cell population that does not display the characteristic tissue architecture of the source organ. In addition, such models do not account for the many interactions between pathogens and the diverse cell subsets that are normally present in a given organ. Conclusions based on conventional 2D cell culture methods are limited, requiring extrapolation from a reductionist model to understand in vivo events. In vitro organ culture (IVOC) offers a way to overcome some of these limitations. Explants conserve important in vivo characteristics, such as different cell types and complex tissue architecture. This in vitro (ex vivo) organ culture protocol of the swine large intestine aims at maintaining viable colonic mucosa for up to 5 days. The protocol described herein applies a combination of methods used for immortalized cell culture and stem cell stimulation to support the physiological cellular flow inherent of the intestinal mucosa. Required equipment includes a hyperoxic chamber and culture at the air-liquid interface.
Collapse
Affiliation(s)
- Matheus de Oliveira Costa
- Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
- Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Michael K Dame
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
2
|
Tian CM, Yang MF, Xu HM, Zhu MZ, Yue NN, Zhang Y, Shi RY, Yao J, Wang LS, Liang YJ, Li DF. Stem cell-derived intestinal organoids: a novel modality for IBD. Cell Death Discov 2023; 9:255. [PMID: 37479716 PMCID: PMC10362068 DOI: 10.1038/s41420-023-01556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023] Open
Abstract
The organoids represent one of the greatest revolutions in the biomedical field in the past decade. This three-dimensional (3D) micro-organ cultured in vitro has a structure highly similar to that of the tissue and organ. Using the regeneration ability of stem cells, a 3D organ-like structure called intestinal organoids is established, which can mimic the characteristics of real intestinal organs, including morphology, function, and personalized response to specific stimuli. Here, we discuss current stem cell-based organ-like 3D intestinal models, including understanding the molecular pathophysiology, high-throughput screening drugs, drug efficacy testing, toxicological evaluation, and organ-based regeneration of inflammatory bowel disease (IBD). We summarize the advances and limitations of the state-of-the-art reconstruction platforms for intestinal organoids. The challenges, advantages, and prospects of intestinal organs as an in vitro model system for precision medicine are also discussed. Key applications of stem cell-derived intestinal organoids. Intestinal organoids can be used to model infectious diseases, develop new treatments, drug screens, precision medicine, and regenerative medicine.
Collapse
Affiliation(s)
- Cheng-Mei Tian
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China
- Department of Emergency, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 51000, China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 51000, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People's Hospital The Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, 516000, Guangdong, China
| | - Rui-Yue Shi
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, 518020, Guangdong, China.
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
3
|
Sena F, Cancela S, Bollati-Fogolín M, Pagotto R, Francia ME. Exploring Toxoplasma gondii´s Biology within the Intestinal Epithelium: intestinal-derived models to unravel sexual differentiation. Front Cell Infect Microbiol 2023; 13:1134471. [PMID: 37313339 PMCID: PMC10258352 DOI: 10.3389/fcimb.2023.1134471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/25/2023] [Indexed: 06/15/2023] Open
Abstract
A variety of intestinal-derived culture systems have been developed to mimic in vivo cell behavior and organization, incorporating different tissue and microenvironmental elements. Great insight into the biology of the causative agent of toxoplasmosis, Toxoplasma gondii, has been attained by using diverse in vitro cellular models. Nonetheless, there are still processes key to its transmission and persistence which remain to be elucidated, such as the mechanisms underlying its systemic dissemination and sexual differentiation both of which occur at the intestinal level. Because this event occurs in a complex and specific cellular environment (the intestine upon ingestion of infective forms, and the feline intestine, respectively), traditional reductionist in vitro cellular models fail to recreate conditions resembling in vivo physiology. The development of new biomaterials and the advances in cell culture knowledge have opened the door to a next generation of more physiologically relevant cellular models. Among them, organoids have become a valuable tool for unmasking the underlying mechanism involved in T. gondii sexual differentiation. Murine-derived intestinal organoids mimicking the biochemistry of the feline intestine have allowed the generation of pre-sexual and sexual stages of T. gondii for the first time in vitro, opening a window of opportunity to tackling these stages by "felinizing" a wide variety of animal cell cultures. Here, we reviewed intestinal in vitro and ex vivo models and discussed their strengths and limitations in the context of a quest for faithful models to in vitro emulate the biology of the enteric stages of T. gondii.
Collapse
Affiliation(s)
- Florencia Sena
- Laboratory of Apicomplexan Biology, Institut Pasteur Montevideo, Montevideo, Uruguay
- Laboratorio de Bioquímica, Departamento de Biología Vegetal, Universidad de la República, Montevideo, Uruguay
| | - Saira Cancela
- Cell Biology Unit, Institut Pasteur Montevideo, Montevideo, Uruguay
- Molecular, Cellular, and Animal Technology Program (ProTeMCA), Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Mariela Bollati-Fogolín
- Cell Biology Unit, Institut Pasteur Montevideo, Montevideo, Uruguay
- Molecular, Cellular, and Animal Technology Program (ProTeMCA), Institut Pasteur Montevideo, Montevideo, Uruguay
| | - Romina Pagotto
- Cell Biology Unit, Institut Pasteur Montevideo, Montevideo, Uruguay
| | - María E. Francia
- Laboratory of Apicomplexan Biology, Institut Pasteur Montevideo, Montevideo, Uruguay
- Departamento de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
4
|
Cortez BRDS, Guedes RMC. A review on the evolution of methods for intestinal in vitro organ culture and its application in veterinary science. Vet World 2023; 16:347-356. [PMID: 37042004 PMCID: PMC10082705 DOI: 10.14202/vetworld.2023.347-356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/06/2023] [Indexed: 02/25/2023] Open
Abstract
Different techniques have been reported in studies of intestinal in vitro organ culture (IVOC). A robust compilation of all available methods is lacking in the literature, making it difficult to choose a method that corresponds to the study's demands. In this review, readers can assess the most available methods, allowing them to evaluate which is more suitable for their purposes and requirements. A simplified view of culturing intestinal explants is presented, highlighting the approachability of IVOC. Relevant findings from diverse veterinarian studies, where explants played a major role, as well as the technique used in each, are described to illustrate its applications. Finally, the strengths and limitations of the innovative intestinal IVOC methods are discussed. This review provides a collection of methods for intestinal explant culture and their possible applications in veterinary research. In this way, it aims to broaden access to IVOC techniques and aid decision-making regarding the best suited for a study's purposes.
Collapse
Affiliation(s)
- Barbara Ribeiro de Souza Cortez
- Department of Veterinary Clinic and Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Large Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Canada
| | | |
Collapse
|
5
|
Hypo-osmotic stress induces the epithelial alarmin IL-33 in the colonic barrier of ulcerative colitis. Sci Rep 2022; 12:11550. [PMID: 35798804 PMCID: PMC9263100 DOI: 10.1038/s41598-022-15573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022] Open
Abstract
Epithelial alarmins are gaining interest as therapeutic targets for chronic inflammation. The nuclear alarmin interleukin-33 (IL-33) is upregulated in the colonic mucosa of acute ulcerative colitis (UC) and may represent an early instigator of the inflammatory cascade. However, it is not clear what signals drive the expression of IL-33 in the colonic mucosa, nor is the exact role of IL-33 elucidated. We established an ex vivo model using endoscopic colonic biopsies from healthy controls and UC patients. Colonic biopsies exposed to hypo-osmotic medium induced a strong nuclear IL-33 expression in colonic crypts in both healthy controls and UC biopsies. Mucosal IL33 mRNA was also significantly increased following hypo-osmotic stress in healthy controls compared to non-stimulated biopsies (fold change 3.9, p-value < 0.02). We observed a modest induction of IL-33 in response to TGF-beta-1 stimulation, whereas responsiveness to inflammatory cytokines TNF and IFN-gamma was negligible. In conclusion our findings indicate that epithelial IL-33 is induced by hypo-osmotic stress, rather than prototypic proinflammatory cytokines in colonic ex vivo biopsies. This is a novel finding, linking a potent cytokine and alarmin of the innate immune system with cellular stress mechanisms and mucosal inflammation.
Collapse
|
6
|
Nguyen DT, Famiglietti JE, Smolchek RA, Dupee Z, Diodati N, Pedro DI, Urueña JM, Schaller MA, Sawyer WG. 3D In Vitro Platform for Cell and Explant Culture in Liquid-like Solids. Cells 2022; 11:cells11060967. [PMID: 35326418 PMCID: PMC8946834 DOI: 10.3390/cells11060967] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Existing 3D cell models and technologies have offered tools to elevate cell culture to a more physiologically relevant dimension. One mechanism to maintain cells cultured in 3D is by means of perfusion. However, existing perfusion technologies for cell culture require complex electronic components, intricate tubing networks, or specific laboratory protocols for each application. We have developed a cell culture platform that simply employs a pump-free suction device to enable controlled perfusion of cell culture media through a bed of granular microgels and removal of cell-secreted metabolic waste. We demonstrated the versatile application of the platform by culturing single cells and keeping tissue microexplants viable for an extended period. The human cardiomyocyte AC16 cell line cultured in our platform revealed rapid cellular spheroid formation after 48 h and ~90% viability by day 7. Notably, we were able to culture gut microexplants for more than 2 weeks as demonstrated by immunofluorescent viability assay and prolonged contractility.
Collapse
Affiliation(s)
- Duy T. Nguyen
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
| | - Jack E. Famiglietti
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
| | - Ryan A. Smolchek
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
| | - Zadia Dupee
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL 32611, USA; (Z.D.); (M.A.S.)
| | - Nickolas Diodati
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
| | - Diego I. Pedro
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
| | - Juan M. Urueña
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
| | - Matthew A. Schaller
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL 32611, USA; (Z.D.); (M.A.S.)
| | - W. Gregory Sawyer
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (D.T.N.); (J.E.F.); (R.A.S.); (N.D.); (D.I.P.); (J.M.U.)
- Correspondence:
| |
Collapse
|
7
|
Elzinga J, van der Lugt B, Belzer C, Steegenga WT. Characterization of increased mucus production of HT29-MTX-E12 cells grown under Semi-Wet interface with Mechanical Stimulation. PLoS One 2021; 16:e0261191. [PMID: 34928974 PMCID: PMC8687553 DOI: 10.1371/journal.pone.0261191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/24/2021] [Indexed: 12/29/2022] Open
Abstract
The intestinal mucus layer plays a crucial role in human health. To study intestinal mucus function and structure in vitro, the mucus-producing intestinal cell line HT29-MTX-E12 has been commonly used. However, this cell line produces only low amounts of the intestine-specific MUC2. It has been shown previously that HT29-MTX-E12 cells cultured under Semi-Wet interface with Mechanical Stimulation (SWMS) produced higher amounts of MUC2, concomitant with a thicker mucus layer, compared to cells cultured conventionally. However, it remains unknown which underlying pathways are involved. Therefore, we aimed to further explore the cellular processes underlying the increased MUC2 production by HT29-MTX-E12 cells grown under SWMS conditions. Cells grown on Transwell membranes for 14 days under static and SWMS conditions (after cell seeding and attachment) were subjected to transcriptome analysis to investigate underlying molecular pathways at gene expression level. Caco-2 and LS174T cell lines were included as references. We characterized how SWMS conditions affected HT29-MTX-E12 cells in terms of epithelial barrier integrity, by measuring transepithelial electrical resistance, and cell metabolism, by monitoring pH and lactate production per molecule glucose of the conditioned medium. We confirmed higher MUC2 production under SWMS conditions at gene and protein level and demonstrated that this culturing method primarily stimulated cell growth. In addition, we also found evidence for a more aerobic cell metabolism under SWMS, as shown previously for similar models. In summary, we suggest different mechanisms by which MUC2 production is enhanced under SWMS and propose potential applications of this model in future studies.
Collapse
Affiliation(s)
- Janneke Elzinga
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Benthe van der Lugt
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Wilma T Steegenga
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
8
|
Glowacki RWP, Engelhart MJ, Ahern PP. Controlled Complexity: Optimized Systems to Study the Role of the Gut Microbiome in Host Physiology. Front Microbiol 2021; 12:735562. [PMID: 34646255 PMCID: PMC8503645 DOI: 10.3389/fmicb.2021.735562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
The profound impact of the gut microbiome on host health has led to a revolution in biomedical research, motivating researchers from disparate fields to define the specific molecular mechanisms that mediate host-beneficial effects. The advent of genomic technologies allied to the use of model microbiomes in gnotobiotic mouse models has transformed our understanding of intestinal microbial ecology and the impact of the microbiome on the host. However, despite incredible advances, our understanding of the host-microbiome dialogue that shapes host physiology is still in its infancy. Progress has been limited by challenges associated with developing model systems that are both tractable enough to provide key mechanistic insights while also reflecting the enormous complexity of the gut ecosystem. Simplified model microbiomes have facilitated detailed interrogation of transcriptional and metabolic functions of the microbiome but do not recapitulate the interactions seen in complex communities. Conversely, intact complex communities from mice or humans provide a more physiologically relevant community type, but can limit our ability to uncover high-resolution insights into microbiome function. Moreover, complex microbiomes from lab-derived mice or humans often do not readily imprint human-like phenotypes. Therefore, improved model microbiomes that are highly defined and tractable, but that more accurately recapitulate human microbiome-induced phenotypic variation are required to improve understanding of fundamental processes governing host-microbiome mutualism. This improved understanding will enhance the translational relevance of studies that address how the microbiome promotes host health and influences disease states. Microbial exposures in wild mice, both symbiotic and infectious in nature, have recently been established to more readily recapitulate human-like phenotypes. The development of synthetic model communities from such "wild mice" therefore represents an attractive strategy to overcome the limitations of current approaches. Advances in microbial culturing approaches that allow for the generation of large and diverse libraries of isolates, coupled to ever more affordable large-scale genomic sequencing, mean that we are now ideally positioned to develop such systems. Furthermore, the development of sophisticated in vitro systems is allowing for detailed insights into host-microbiome interactions to be obtained. Here we discuss the need to leverage such approaches and highlight key challenges that remain to be addressed.
Collapse
Affiliation(s)
- Robert W. P. Glowacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Morgan J. Engelhart
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Philip P. Ahern
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
9
|
De Angelis M, Siragusa S, Vacca M, Di Cagno R, Cristofori F, Schwarm M, Pelzer S, Flügel M, Speckmann B, Francavilla R, Gobbetti M. Selection of Gut-Resistant Bacteria and Construction of Microbial Consortia for Improving Gluten Digestion under Simulated Gastrointestinal Conditions. Nutrients 2021; 13:nu13030992. [PMID: 33808622 PMCID: PMC8003469 DOI: 10.3390/nu13030992] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
This work aimed to define the microbial consortia that are able to digest gluten into non-toxic and non-immunogenic peptides in the human gastrointestinal tract. Methods: 131 out of 504 tested Bacillus and lactic acid bacteria, specifically Bacillus (64), lactobacilli (63), Pediococcus (1), and Weissella (3), showed strong gastrointestinal resistance and were selected for their PepN, PepI, PepX, PepO, and PepP activities toward synthetic substrates. Based on multivariate analysis, 24 strains were clearly distinct from the other tested strains based on having the highest enzymatic activities. As estimated by RP-HPLC and nano-ESI–MS/MS, 6 cytoplasmic extracts out of 24 selected strains showed the ability to hydrolyze immunogenic epitopes, specifically 57–68 of α9-gliadin, 62–75 of A-gliadin, 134–153 of γ-gliadin, and 57–89 (33-mer) of α2-gliadin. Live and lysed cells of selected strains were combined into different microbial consortia for hydrolyzing gluten under gastrointestinal conditions. Commercial proteolytic enzymes (Aspergillusoryzae E1, Aspergillusniger E2, Bacillussubtilis Veron HPP, and Veron PS proteases) were also added to each microbial consortium. Consortium activity was evaluated by ELISA tests, RP-HPLC-nano-ESI–MS/MS, and duodenal explants from celiac disease patients. Results: two microbial consortia (Consortium 4: Lactiplantibacillus (Lp.) plantarum DSM33363 and DSM33364, Lacticaseibacillus (Lc.) paracasei DSM33373, Bacillussubtilis DSM33298, and Bacilluspumilus DSM33301; and Consortium 16: Lp. plantarum DSM33363 and DSM33364, Lc. paracasei DSM33373, Limosilactobacillusreuteri DSM33374, Bacillusmegaterium DSM33300, B.pumilus DSM33297 and DSM33355), containing commercial enzymes, were able to hydrolyze gluten to non-toxic and non-immunogenic peptides under gastrointestinal conditions. Conclusions: the results of this study provide evidence that selected microbial consortia could potentially improve the digestion of gluten in gluten-sensitive patients by hydrolyzing the immunogenic peptides during gastrointestinal digestion.
Collapse
Affiliation(s)
- Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.D.A.); (S.S.); (M.V.)
| | - Sonya Siragusa
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.D.A.); (S.S.); (M.V.)
| | - Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.D.A.); (S.S.); (M.V.)
| | - Raffaella Di Cagno
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| | - Fernanda Cristofori
- Interdisciplinary Department of Medicine-Pediatric Section, University of Bari Aldo Moro, Ospedale Pediatrico Giovanni XXIII, 70125 Bari, Italy; (F.C.); (R.F.)
| | - Michael Schwarm
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Stefan Pelzer
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Monika Flügel
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Bodo Speckmann
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine-Pediatric Section, University of Bari Aldo Moro, Ospedale Pediatrico Giovanni XXIII, 70125 Bari, Italy; (F.C.); (R.F.)
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
- Correspondence: ; Tel.: +39-0471-017215
| |
Collapse
|
10
|
Coleman OI, Haller D. Microbe-Mucus Interface in the Pathogenesis of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13040616. [PMID: 33557139 PMCID: PMC7913824 DOI: 10.3390/cancers13040616] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
Overlying gastrointestinal epithelial cells is the transparent mucus layer that separates the lumen from the host. The dynamic mucus layer serves to lubricate the mucosal surface, to protect underlying epithelial cells, and as a transport medium between luminal contents and epithelial cells. Furthermore, it provides a habitat for commensal bacteria and signals to the underlying immune system. Mucins are highly glycosylated proteins, and their glycocode is tissue-specific and closely linked to the resident microbiota. Aberrant mucin expression and glycosylation are linked to chronic inflammation and gastrointestinal cancers, including colorectal cancer (CRC). Aberrant mucus production compromises the mucus layer and allows bacteria to come into close contact with the intestinal epithelium, potentially triggering unfavorable host responses and the subsequent development of tumors. Here, we review our current understanding of the interaction between the intestinal microbiota and mucus in healthy and CRC subjects. Deep knowledge of the intricate mechanisms of microbe-mucus interactions may contribute to the development of novel treatment strategies for CRC, in which a dysfunctional mucus layer is observed.
Collapse
Affiliation(s)
- Olivia I. Coleman
- Department of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany;
- Correspondence: ; Tel.: +49-08161-71-2375
| | - Dirk Haller
- Department of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany;
- ZIEL—Institute for Food & Health, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
11
|
Chirdo FG, Auricchio S, Troncone R, Barone MV. The gliadin p31-43 peptide: Inducer of multiple proinflammatory effects. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 358:165-205. [PMID: 33707054 DOI: 10.1016/bs.ircmb.2020.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coeliac disease (CD) is the prototype of an inflammatory chronic disease induced by food. In this context, gliadin p31-43 peptide comes into the spotlight as an important player of the inflammatory/innate immune response to gliadin in CD. The p31-43 peptide is part of the p31-55 peptide from α-gliadins that remains undigested for a long time, and can be present in the small intestine after ingestion of a gluten-containing diet. Different biophysical methods and molecular dynamic simulations have shown that p31-43 spontaneously forms oligomeric nanostructures, whereas experimental approaches using in vitro assays, mouse models, and human duodenal tissues have shown that p31-43 is able to induce different forms of cellular stress by driving multiple inflammatory pathways. Increased proliferative activity of the epithelial cells in the crypts, enterocyte stress, activation of TG2, induction of Ca2+, IL-15, and NFκB signaling, inhibition of CFTR, alteration of vesicular trafficking, and activation of the inflammasome platform are some of the biological effects of p31-43, which, in the presence of appropriate genetic susceptibility and environmental factors, may act together to drive CD.
Collapse
Affiliation(s)
- Fernando Gabriel Chirdo
- Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Instituto de Estudios Inmunológicos y Fisiopatológicos-IIFP (UNLP-CONICET), La Plata, Argentina.
| | - Salvatore Auricchio
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University Federico II, Naples, Italy
| | - Riccardo Troncone
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University Federico II, Naples, Italy; Department of Translational Medical Science, University Federico II, Naples, Italy
| | - Maria Vittoria Barone
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University Federico II, Naples, Italy; Department of Translational Medical Science, University Federico II, Naples, Italy
| |
Collapse
|
12
|
Mayorgas A, Dotti I, Salas A. Microbial Metabolites, Postbiotics, and Intestinal Epithelial Function. Mol Nutr Food Res 2020; 65:e2000188. [DOI: 10.1002/mnfr.202000188] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/31/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Aida Mayorgas
- Department of Gastroenterology, Hospital Clínic ‐ IDIBAPS C/Rosselló, 149‐153, 3rd Floor Barcelona 08036 Spain
| | - Isabella Dotti
- Department of Gastroenterology, Hospital Clínic ‐ IDIBAPS C/Rosselló, 149‐153, 3rd Floor Barcelona 08036 Spain
| | - Azucena Salas
- Department of Gastroenterology, Hospital Clínic ‐ IDIBAPS C/Rosselló, 149‐153, 3rd Floor Barcelona 08036 Spain
| |
Collapse
|
13
|
Li AP. In Vitro Human Cell-Based Experimental Models for the Evaluation of Enteric Metabolism and Drug Interaction Potential of Drugs and Natural Products. Drug Metab Dispos 2020; 48:980-992. [PMID: 32636209 DOI: 10.1124/dmd.120.000053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/18/2020] [Indexed: 02/13/2025] Open
Abstract
Elements of key enteric drug metabolism and disposition pathways are reviewed to aid the assessment of the applicability of current cell-based enteric experimental systems for the evaluation of enteric metabolism and drug interaction potential. Enteric nuclear receptors include vitamin D receptor, constitutive androstane receptor, pregnane X receptor, farnesoid X receptor, liver X receptor, aryl hydrocarbon receptor, and peroxisome proliferator-activated receptor. Enteric drug metabolizing enzyme pathways include both cytochrome P450 (P450) and non-P450 drug metabolizing enzymes based on gene expression, proteomics, and activity. Both uptake and efflux transporters are present in the small intestine, with P-glycoprotein found to be responsible for most drug-drug and food-drug interactions. The cell-based in vitro enteric systems reviewed are 1) immortalized cell line model: the human colon adenocarcinoma (Caco-2) cells; 2) human stem cell-derived enterocyte models: stem cell enteric systems, either from intestinal crypt cells or induced pluripotent stem cells; and 3) primary cell models: human intestinal slices, cryopreserved human enterocytes, permeabilized cofactor-supplemented (MetMax) cryopreserved human enterocytes, and cryopreserved human intestinal mucosa. The major deficiency with both immortalized cell lines and stem cell-derived enterocytes is that drug metabolizing enzyme activities, although they are detectable, are substantially lower than those for the intestinal mucosa in vivo. Human intestine slices, cryopreserved human enterocytes, MetMax cryopreserved human enterocytes, and cryopreserved human intestinal mucosa retain robust enteric drug metabolizing enzyme activity and represent appropriate models for the evaluation of metabolism and metabolism-dependent drug interaction potential of orally administered xenobiotics including drugs, botanical products, and dietary supplements. SIGNIFICANCE STATEMENT: Enteric drug metabolism plays an important role in the bioavailability and metabolic fate of orally administered drugs as well as in enteric drug-drug and food-drug interactions. The current status of key enteric drug metabolism and disposition pathways and in vitro human cell-based enteric experimental systems for the evaluation of the metabolism and drug interaction potential of orally administered substances is reviewed.
Collapse
Affiliation(s)
- Albert P Li
- In Vitro ADMET Laboratories, Inc., Columbia, Maryland
| |
Collapse
|
14
|
Danielsen EM. Intestinal permeation enhancers: Lessons learned from studies using an organ culture model. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183474. [PMID: 32946886 DOI: 10.1016/j.bbamem.2020.183474] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/18/2020] [Accepted: 09/09/2020] [Indexed: 01/18/2023]
Abstract
Permeation enhancers (PEs) are compounds aimed to increase intestinal uptake of oral drugs with poor bioavailability. This mini-review focuses on results recently obtained with PEs using an intestinal organ culture model. The model predicts which paracellular/transcellular pathways across the epithelium are susceptible to different classes of PEs (mainly surfactants and cell penetrating peptides). PEs: 1) generate a transmembrane transcellular pathway, 2) block apical endocytosis (first step in apical-to-basolateral transcytosis), and 3) perturb normal cell membrane integrity. The results argue that surfactants and cell penetrating peptides are not suitable for use in formulations aimed to exploit transcytosis in oral drug delivery.
Collapse
Affiliation(s)
- E Michael Danielsen
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
15
|
Kramer N, Pratscher B, Meneses AMC, Tschulenk W, Walter I, Swoboda A, Kruitwagen HS, Schneeberger K, Penning LC, Spee B, Kieslinger M, Brandt S, Burgener IA. Generation of Differentiating and Long-Living Intestinal Organoids Reflecting the Cellular Diversity of Canine Intestine. Cells 2020; 9:cells9040822. [PMID: 32231153 PMCID: PMC7226743 DOI: 10.3390/cells9040822] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/23/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Functional intestinal disorders constitute major, potentially lethal health problems in humans. Consequently, research focuses on elucidating the underlying pathobiological mechanisms and establishing therapeutic strategies. In this context, intestinal organoids have emerged as a potent in vitro model as they faithfully recapitulate the structure and function of the intestinal segment they represent. Interestingly, human-like intestinal diseases also affect dogs, making canine intestinal organoids a promising tool for canine and comparative research. Therefore, we generated organoids from canine duodenum, jejunum and colon, and focused on simultaneous long-term expansion and cell differentiation to maximize applicability. Following their establishment, canine intestinal organoids were grown under various culture conditions and then analyzed with respect to cell viability/apoptosis and multi-lineage differentiation by transcription profiling, proliferation assay, cell staining, and transmission electron microscopy. Standard expansion medium supported long-term expansion of organoids irrespective of their origin, but inhibited cell differentiation. Conversely, transfer of organoids to differentiation medium promoted goblet cell and enteroendocrine cell development, but simultaneously induced apoptosis. Unimpeded stem cell renewal and concurrent differentiation was achieved by culturing organoids in the presence of tyrosine kinase ligands. Our findings unambiguously highlight the characteristic cellular diversity of canine duodenum, jejunum and colon as fundamental prerequisite for accurate in vitro modelling.
Collapse
Affiliation(s)
- Nina Kramer
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
- Correspondence:
| | - Barbara Pratscher
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Andre M. C. Meneses
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | - Waltraud Tschulenk
- Institute of Pathology, Department for Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Ingrid Walter
- Institute of Pathology, Department for Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Alexander Swoboda
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Hedwig S. Kruitwagen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | - Louis C. Penning
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, The Netherlands
| | - Matthias Kieslinger
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Sabine Brandt
- Research Group Oncology, Equine Surgery, Department of Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Iwan A. Burgener
- Division of Small Animal Internal Medicine, Department for Small Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
16
|
Danielsen EM, De Haro Hernando A, Yassin M, Rasmussen K, Olsen J, Hansen GH, Danielsen EM. Short-term tissue permeability actions of dextran sulfate sodium studied in a colon organ culture system. Tissue Barriers 2020; 8:1728165. [PMID: 32079482 PMCID: PMC7549740 DOI: 10.1080/21688370.2020.1728165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dextran sulfate sodium (DSS)-induced colitis is the most commonly used animal model for inflammatory bowel diseases. However, the precise molecular action of DSS, in particular its initial effect on the epithelial tissue permeability, is still poorly understood. In the present work, organ culture of mouse – and pig colon explants were performed for 1–2 h in the presence/absence of 2% DSS together with polar- and lipophilic fluorescent probes. Probe permeability was subsequently assessed by fluorescence microscopy. DSS rapidly increased paracellular permeability of 70-kDa dextran without otherwise affecting the overall epithelial integrity. FITC-conjugated DSS likewise permeated the epithelial barrier and strongly accumulated in nuclei of cells scattered in the lamina propria. By immunolabeling, plasma cells, T cells, macrophages, mast cells, and fibroblasts were identified as possible targets for DSS, indicating that accumulation of the polyanion in nuclei was not confined to a particular type of cell in the lamina propria. In contrast, colonocytes were rarely targeted by DSS, but as visualized by transmission electron microscopy, it induced the formation of vacuole-like structures in the intercellular space between adjacent epithelial cells. Nuclei of various cell types in the lamina propria, including both cells of the innate and adaptive immune system, are novel targets for a rapid action of DSS, and from previous in vitro studies, polyanions like DSS are known to disrupt nucleosomes by binding to the histones. We therefore propose that nuclear targeting is one way whereby DSS exerts its inflammatory action as a colitogen in animal models of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Elisabeth M Danielsen
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Alba De Haro Hernando
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Mohammad Yassin
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Karina Rasmussen
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Jørgen Olsen
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Gert H Hansen
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| | - E Michael Danielsen
- Department of Cellular and Molecular Medicine, the Panum Institute, Faculty of Health Sciences, University of Copenhagen , Copenhagen, Denmark
| |
Collapse
|
17
|
Wang L, Li Y, Guo B, Zhang J, Zhu B, Li H, Ding Y, Meng B, Zhao H, Xiang L, Dong J, Liu M, Zhang J, Xiang L, Xiang G. Myeloid-Derived Growth Factor Promotes Intestinal Glucagon-Like Peptide-1 Production in Male Mice With Type 2 Diabetes. Endocrinology 2020; 161:5698328. [PMID: 31913472 DOI: 10.1210/endocr/bqaa003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022]
Abstract
Myeloid-derived growth factor (MYDGF), which is produced by bone marrow-derived cells, mediates cardiac repair following myocardial infarction by inhibiting cardiac myocyte apoptosis to subsequently reduce the infarct size. However, the function of MYDGF in the incretin system of diabetes is still unknown. Here, loss-of-function and gain-of-function experiments in mice revealed that MYDGF maintains glucose homeostasis by inducing glucagon-like peptide-1 (GLP-1) production and secretion and that it improves glucose tolerance and lipid metabolism. Treatment with recombinant MYDGF increased the secretion and production of GLP-1 in STC-1 cells in vitro. Mechanistically, the positive effects of MYDGF are potentially attributable to the activation of protein kinase A/glycogen synthase kinase 3β/β-catenin (PKA/GSK-3β/β-catenin) and mitogen-activated protein kinase (MAPK) kinases/extracellular regulated protein kinase (MEK/ERK) pathways. Based on these findings, MYDGF promotes the secretion and production of GLP-1 in intestinal L-cells and potentially represents a potential therapeutic medication target for type 2 diabetes.
Collapse
Affiliation(s)
- Li Wang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Bei Guo
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiajia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Biao Zhu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Huan Li
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Yan Ding
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Biying Meng
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zhao
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Lin Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | - Junxia Zhang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
| | | | - Guangda Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuhan, Hubei Province, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
18
|
Baydoun M, Treizeibré A, Follet J, Benamrouz Vanneste S, Creusy C, Dercourt L, Delaire B, Mouray A, Viscogliosi E, Certad G, Senez V. An Interphase Microfluidic Culture System for the Study of Ex Vivo Intestinal Tissue. MICROMACHINES 2020; 11:E150. [PMID: 32019215 PMCID: PMC7074597 DOI: 10.3390/mi11020150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/21/2020] [Accepted: 01/28/2020] [Indexed: 11/22/2022]
Abstract
Ex vivo explant culture models offer unique properties to study complex mechanisms underlying tissue growth, renewal, and disease. A major weakness is the short viability depending on the biopsy origin and preparation protocol. We describe an interphase microfluidic culture system to cultivate full thickness murine colon explants which keeps morphological structures of the tissue up to 192 h. The system was composed of a central well on top of a porous membrane supported by a microchannel structure. The microfluidic perfusion allowed bathing the serosal side while preventing immersion of the villi. After eight days, up to 33% of the samples displayed no histological abnormalities. Numerical simulation of the transport of oxygen and glucose provided technical solutions to improve the functionality of the microdevice.
Collapse
Affiliation(s)
- Martha Baydoun
- Univ. Lille, CNRS, ISEN-YNCREA, UMR 8520-IEMN, F-59000 Lille, France
- ISA-YNCREA Hauts de France, F-59000 Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9107-CIIL-Centre d’Infection et d’Immunité de Lille, F-59019 Lille, France
| | | | - Jérôme Follet
- Univ. Lille, CNRS, ISEN-YNCREA, UMR 8520-IEMN, F-59000 Lille, France
- ISA-YNCREA Hauts de France, F-59000 Lille, France
| | - Sadia Benamrouz Vanneste
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9107-CIIL-Centre d’Infection et d’Immunité de Lille, F-59019 Lille, France
- Laboratoire Ecologie et Biodiversité, Unité de Recherche Smart and Sustainable Cities, Faculté de Gestion Economie et Sciences, Institut Catholique de Lille, F-59800 Lille, France
| | - Colette Creusy
- Service d’Anatomie et de Cytologie Pathologiques, Groupement des Hôpitaux de l’Université Catholique de Lille, 59000 Lille, France
| | - Lucie Dercourt
- CNRS, Univ. Tokyo, UMI 2820 — LIMMS, F-59000 Lille, France
| | - Baptiste Delaire
- Service d’Anatomie et de Cytologie Pathologiques, Groupement des Hôpitaux de l’Université Catholique de Lille, 59000 Lille, France
| | - Anthony Mouray
- Plateforme d’Expérimentations et de Hautes Technologies Animales, Institut Pasteur de Lille Lille, 59019 Lille, France
| | - Eric Viscogliosi
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9107-CIIL-Centre d’Infection et d’Immunité de Lille, F-59019 Lille, France
| | - Gabriela Certad
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9107-CIIL-Centre d’Infection et d’Immunité de Lille, F-59019 Lille, France
- Délégation à la Recherche Clinique et à l’Innovation, Groupement des Hôpitaux de l’Institut Catholique de Lille (GHICL), Faculté de Médecine et Maïeutique, Université Catholique de Lille, 59800 Lille, France
| | - Vincent Senez
- Univ. Lille, CNRS, ISEN-YNCREA, UMR 8520-IEMN, F-59000 Lille, France
- CNRS, Univ. Tokyo, UMI 2820 — LIMMS, F-59000 Lille, France
| |
Collapse
|
19
|
Etienne-Mesmin L, Chassaing B, Desvaux M, De Paepe K, Gresse R, Sauvaitre T, Forano E, de Wiele TV, Schüller S, Juge N, Blanquet-Diot S. Experimental models to study intestinal microbes–mucus interactions in health and disease. FEMS Microbiol Rev 2019; 43:457-489. [DOI: 10.1093/femsre/fuz013] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
ABSTRACT
A close symbiotic relationship exists between the intestinal microbiota and its host. A critical component of gut homeostasis is the presence of a mucus layer covering the gastrointestinal tract. Mucus is a viscoelastic gel at the interface between the luminal content and the host tissue that provides a habitat to the gut microbiota and protects the intestinal epithelium. The review starts by setting up the biological context underpinning the need for experimental models to study gut bacteria-mucus interactions in the digestive environment. We provide an overview of the structure and function of intestinal mucus and mucins, their interactions with intestinal bacteria (including commensal, probiotics and pathogenic microorganisms) and their role in modulating health and disease states. We then describe the characteristics and potentials of experimental models currently available to study the mechanisms underpinning the interaction of mucus with gut microbes, including in vitro, ex vivo and in vivo models. We then discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Benoit Chassaing
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303 , USA
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303 , USA
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Kim De Paepe
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Raphaële Gresse
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Thomas Sauvaitre
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Evelyne Forano
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR7UQ, United Kingdom
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRA, MEDIS, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
20
|
Abstract
Models have been extensively used to investigate disease pathogenesis. Animal models are costly, require extensive logistics for animal care, and samples are not always suitable for different analytical techniques or to answer the research question. In vitro cell culture models are generally focused on recreating a specific characteristic of an organ, and are limited to a single cell population that does not display the characteristic tissue architecture of the source organ. In addition, such models do not account for the many interactions between pathogens and the diverse cell subsets that are normally present in a given organ. Conclusions based on conventional 2D cell culture methods are limited, requiring extrapolation from a reductionist model to understand in vivo events. In vitro organ culture (IVOC) offers a way to overcome some of these limitations. Explants conserve important in vivo characteristics, such as different cell types and complex tissue architecture. This in vitro (ex vivo) organ culture protocol of the swine large intestine aims at maintaining viable colonic mucosa for up to 5 days. The protocol described herein applies a combination of methods used for immortalized cell culture and stem cell stimulation to support the physiological cellular flow inherent of the intestinal mucosa. Required equipment includes a hyperoxic chamber and culture at the air-liquid interface.
Collapse
Affiliation(s)
- Matheus O Costa
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada. .,Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Janet E Hill
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Michael K Dame
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, MI, USA
| | - John C S Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
21
|
DA COSTA GONÇALVES FABIANY, SERAFINI MICHELEARAMBURU, MELLO HELENAFLORES, PFAFFENSELLER BIANCA, ARAÚJO ANELISEBERGMANN, VISIOLI FERNANDA, PAZ ANAHELENA. Bioactive factors secreted from mesenchymal stromal cells protect the intestines from experimental colitis in a three-dimensional culture. Cytotherapy 2018; 20:1459-1471. [PMID: 30523788 DOI: 10.1016/j.jcyt.2018.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/24/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023]
|
22
|
Pearce SC, Coia HG, Karl JP, Pantoja-Feliciano IG, Zachos NC, Racicot K. Intestinal in vitro and ex vivo Models to Study Host-Microbiome Interactions and Acute Stressors. Front Physiol 2018; 9:1584. [PMID: 30483150 PMCID: PMC6240795 DOI: 10.3389/fphys.2018.01584] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
The gut microbiome is extremely important for maintaining homeostasis with host intestinal epithelial, neuronal, and immune cells and this host-microbe interaction is critical during times of stress or disease. Environmental, nutritional, and cognitive stress are just a few factors known to influence the gut microbiota and are thought to induce microbial dysbiosis. Research on this bidirectional relationship as it pertains to health and disease is extensive and rapidly expanding in both in vivo and in vitro/ex vivo models. However, far less work has been devoted to studying effects of host-microbe interactions on acute stressors and performance, the underlying mechanisms, and the modulatory effects of different stressors on both the host and the microbiome. Additionally, the use of in vitro/ex vivo models to study the gut microbiome and human performance has not been researched extensively nor reviewed. Therefore, this review aims to examine current evidence concerning the current status of in vitro and ex vivo host models, the impact of acute stressors on gut physiology/microbiota as well as potential impacts on human performance and how we can parlay this information for DoD relevance as well as the broader scientific community. Models reviewed include widely utilized intestinal cell models from human and animal models that have been applied in the past for stress or microbiology research as well as ex vivo organ/tissue culture models and new innovative models including organ-on-a-chip and co-culture models.
Collapse
Affiliation(s)
- Sarah C Pearce
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Heidi G Coia
- National Research Council, The National Academies of Sciences, Engineering, and Medicine, Washington, DC, United States.,711th Human Performance Wing, Airforce Research Laboratory, Airman Systems Directorate, Human-Centered ISR Division, Molecular Mechanisms Branch, Wright-Patterson Air Force Base, Dayton, OH, United States
| | - J P Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Ida G Pantoja-Feliciano
- Soldier Protection and Optimization Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Nicholas C Zachos
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kenneth Racicot
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The medical management of inflammatory bowel disease (IBD) remains problematic with a pressing need for innovation in drug development as well as delivery of personalized therapies. Both the disease's inherent pathophysiologic complexity and heterogeneity in its etiology conspire in making it difficult to accurately model for either the purposes of basic research or drug development. Multiple attempts at creating meaningful experimental models have fallen short of adequately recapitulating the disease and most do not capture any aspect of the cause or the effects of patient heterogeneity that underlays most of the difficulties faced by physicians and their patients. In vivo animal models, tissue culture systems, and more recent synthetic biology approaches are all too simplistically reductionist for the task. However, ex vivo culture platforms utilizing patient biopsies offer a system that more closely mimics end-stage disease processes that can be studied in detail and subjected to experimental manipulations. RECENT FINDINGS Recent studies describe further optimization of mucosal explant cultures in order to increase tissue viability and maintain a polarized epithelial layer. Current applications of the platform include studies of the interplay between the epithelial, immune and stromal compartment of the intestinal tissue, investigation of host-microbial interactions, preclinical evaluation of candidate drugs and uncovering mechanisms of action of established or emerging treatments for IBD. SUMMARY Patient explant-based assays offer an advanced biological system in IBD that recapitulates disease complexity and reflects the heterogeneity of the patient population. In its current stage of development, the system can be utilized for drug testing prior to the costlier and time-consuming evaluation by clinical trials. Further refinement of the technology and establishment of assay readouts that correlate with therapeutic outcomes will yield a powerful tool for personalized medicine approaches in which individual patient responses to available treatments are assessed a priori, thus reducing the need for trial and error within the clinical setting.
Collapse
|
24
|
Abstract
The human gut microbiome performs prodigious physiological functions such as production of microbial metabolites, modulation of nutrient digestion and drug metabolism, control of immune system, and prevention of infection. Paradoxically, gut microbiome can also negatively orchestrate the host responses in diseases or chronic disorders, suggesting that the regulated and balanced host-gut microbiome crosstalk is a salient prerequisite in gastrointestinal physiology. To understand the pathophysiological role of host-microbiome crosstalk, it is critical to recreate in vivo relevant models of the host-gut microbiome ecosystem in human. However, controlling the multi-species microbial communities and their uncontrolled growth has remained a notable technical challenge. Furthermore, conventional two-dimensional (2D) or 3D culture systems do not recapitulate multicellular microarchitectures, mechanical dynamics, and tissue-specific functions. Here, we review recent advances and current pitfalls of in vitro and ex vivo models that display human GI functions. We also discuss how the disruptive technologies such as 3D organoids or a human organ-on-a-chip microphysiological system can contribute to better emulate host-gut microbiome crosstalks in health and disease. Finally, the medical and pharmaceutical significance of the gut microbiome-based personalized interventions is underlined as a future perspective.
Collapse
|
25
|
Maresca M, Pinton P, Ajandouz EH, Menard S, Ferrier L, Oswald IP. Overview and Comparison of Intestinal Organotypic Models, Intestinal Cells, and Intestinal Explants Used for Toxicity Studies. Curr Top Microbiol Immunol 2018; 430:247-264. [PMID: 30259111 DOI: 10.1007/82_2018_142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The intestine is a complex organ formed of different types of cell distributed in different layers of tissue. To minimize animal experiments, for decades, researchers have been trying to develop in vitro/ex vivo systems able to mimic the cellular diversity naturally found in the gut. Such models not only help our understanding of the gut physiology but also of intestinal toxicity. This review describes the different systems used to evaluate the effects of drugs/contaminants on intestinal functions and compares their advantages and limitations. The comparison showed that the organotypic model is the best available model to perform intestinal toxicity studies, including on human tissues.
Collapse
Affiliation(s)
- Marc Maresca
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Philippe Pinton
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Sandrine Menard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Laurent Ferrier
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Isabelle P Oswald
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
26
|
Fernando EH, Dicay M, Stahl M, Gordon MH, Vegso A, Baggio C, Alston L, Lopes F, Baker K, Hirota S, McKay DM, Vallance B, MacNaughton WK. A simple, cost-effective method for generating murine colonic 3D enteroids and 2D monolayers for studies of primary epithelial cell function. Am J Physiol Gastrointest Liver Physiol 2017; 313:G467-G475. [PMID: 28751424 DOI: 10.1152/ajpgi.00152.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 01/31/2023]
Abstract
Cancer cell lines have been the mainstay of intestinal epithelial experimentation for decades, due primarily to their immortality and ease of culture. However, because of the inherent biological abnormalities of cancer cell lines, many cellular biologists are currently transitioning away from these models and toward more representative primary cells. This has been particularly challenging, but recent advances in the generation of intestinal organoids have brought the routine use of primary cells within reach of most epithelial biologists. Nevertheless, even with the proliferation of publications that use primary intestinal epithelial cells, there is still a considerable amount of trial and error required for laboratories to establish a consistent and reliable method to culture three-dimensional (3D) intestinal organoids and primary epithelial monolayers. We aim to minimize the time other laboratories spend troubleshooting the technique and present a standard method for culturing primary epithelial cells. Therefore, we have described our optimized, high-yield, cost-effective protocol to grow 3D murine colonoids for more than 20 passages and our detailed methods to culture these cells as confluent monolayers for at least 14 days, enabling a wide variety of potential future experiments. By supporting and expanding on the current literature of primary epithelial culture optimization and detailed use in experiments, we hope to help enable the widespread adoption of these innovative methods and allow consistency of results obtained across laboratories and institutions.NEW & NOTEWORTHY Primary intestinal epithelial monolayers are notoriously difficult to maintain culture, even with the recent advances in the field. We describe, in detail, the protocols required to maintain three-dimensional cultures of murine colonoids and passage these primary epithelial cells to confluent monolayers in a standardized, high-yield and cost-effective manner.
Collapse
Affiliation(s)
- Elizabeth H Fernando
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Michael Dicay
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Martin Stahl
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Marilyn H Gordon
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Andrew Vegso
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Cristiane Baggio
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Laurie Alston
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Fernando Lopes
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Kristi Baker
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Simon Hirota
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Derek M McKay
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - Bruce Vallance
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Wallace K MacNaughton
- Department of Physiology and Pharmacology and Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada;
| |
Collapse
|
27
|
Vadstrup K, Galsgaard ED, Jensen H, Lanier LL, Ryan JC, Chen SY, Nolan GP, Vester-Andersen MK, Pedersen JS, Gerwien J, Jensen T, Bendtsen F. NKG2D ligand expression in Crohn's disease and NKG2D-dependent stimulation of CD8 + T cell migration. Exp Mol Pathol 2017; 103:56-70. [PMID: 28684217 DOI: 10.1016/j.yexmp.2017.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 05/24/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022]
Abstract
Interaction between the activating NKG2D receptor on lymphocytes and its ligands MICA, MICB, and ULBP1-6 modulate T and NK cell activity and may contribute to the pathogenesis of Crohn's disease (CD). NKG2D ligands are generally not expressed on the cell surface of normal, non-stressed cells, but expression of MICA and MICB in CD intestine has been reported. In this exploratory study, we further characterize the expression of NKG2D and its ligands, including the less well-described ULBP4-6, in CD, and test if NKG2D ligand interactions are involved in the migration of activated T cells into the affected mucosal compartments. Intestinal tissue from CD patients and healthy controls were analyzed by flow cytometry, mass cytometry, and immunohistochemistry for expression of NKG2D and ligands, and for cytokine release. Furthermore, NKG2D-dependent chemotaxis of activated CD8+ T cells across a monolayer of ligand-expressing human intestinal endothelial cells was examined. Activated lymphocytes down-regulated NKG2D expression upon accumulation in inflamed CD intestine. NKG2D expression on CD56+ T and γδ T cells from inflamed tissue seemed inversely correlated with CRP levels and cytokine release. B cells, monocytes, mucosal epithelium, and vascular endothelium expressed NKG2D ligands in inflamed CD intestine. The expression of NKG2D ligands was correlated with cytokine release, but was highly variable between patients. Stimulation of vascular intestinal endothelial cells in vitro induced expression of NKG2D ligands, including MICA/B and ULBP2/6. Blockade of NKG2D on CD8+ T cells inhibited the migration over ligand-expressing endothelial cells. Intestinal induction of NKG2D ligands and ligand-induced down-regulation of NKG2D in CD suggest that the NKG2D-ligand interaction may be involved in both the activation and recruitment of NKG2D+ lymphocytes into the inflamed CD intestine.
Collapse
Affiliation(s)
- Kasper Vadstrup
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark; Faculty of Health Sciences, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark; Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | - Helle Jensen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James C Ryan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, Veterans Affairs Medical Center and University of California San Francisco, San Francisco, CA, USA
| | - Shih-Yu Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | | | - Julie Steen Pedersen
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark
| | - Jens Gerwien
- Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark
| | - Teis Jensen
- Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Maaloev, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Division, Hvidovre University Hospital, DK-2650 Hvidovre, Denmark; Faculty of Health Sciences, The Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
28
|
Selected Probiotic Lactobacilli Have the Capacity To Hydrolyze Gluten Peptides during Simulated Gastrointestinal Digestion. Appl Environ Microbiol 2017; 83:AEM.00376-17. [PMID: 28500039 DOI: 10.1128/aem.00376-17] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/02/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to demonstrate the capacity of probiotic lactobacilli to hydrolyze immunogenic gluten peptides. Eighteen commercial strains of probiotic lactobacilli with highly variable peptidase activity (i.e., aminopeptidase N, iminopeptidase, prolyl endopeptidyl peptidase, tripeptidase, prolidase, prolinase, and dipeptidase), including toward Pro-rich peptides, were tested in this study. Ten probiotic strains were selected on the basis of their specific enzyme activity. When pooled, these 10 strains provided the peptidase portfolio that is required to completely degrade the immunogenic gluten peptides involved in celiac disease (CD). The selected probiotic mixture was able to completely hydrolyze well-known immunogenic epitopes, including the gliadin 33-mer peptide, the peptide spanning residues 57 to 68 of the α9-gliadin (α9-gliadin peptide 57-68), A-gliadin peptide 62-75, and γ-gliadin peptide 62-75. During digestion under simulated gastrointestinal conditions, the pool of 10 selected probiotic lactobacilli strongly hydrolyzed the wheat bread gluten (ca. 18,000 ppm) to less than 10 ppm after 360 min of treatment. As determined by multidimensional chromatography (MDLC) coupled to nanoelectrospray ionization (nano-ESI)-tandem mass spectrometry (MS/MS), no known immunogenic peptides were detected in wheat bread that was digested in the presence of the probiotics. Accordingly, the level of cytokines (interleukin 2 [IL-2], IL-10, and interferon gamma [IFN-γ]) produced by duodenal biopsy specimens from CD patients who consumed wheat bread digested by probiotics was similar to the baseline value (negative control). Probiotics that specifically hydrolyze gluten polypeptides could also be used to hydrolyze immunogenic peptides that contaminate gluten-free products. This could provide a new and safe adjunctive therapy alternative to the gluten-free diet (GFD).IMPORTANCE This study confirmed that probiotic Lactobacillus strains have different enzymatic abilities for hydrolyzing polypeptides, including the Pro-rich epitopes involved in the pathology of CD. Ten lactobacilli with complementary peptidase activities that hydrolyze gluten peptides during simulated gastrointestinal digestion were selected and tested. The results collected showed the potential of probiotic formulas as novel dietary treatments for CD patients.
Collapse
|
29
|
Diagnostic and Research Aspects of Small Intestinal Disaccharidases in Coeliac Disease. J Immunol Res 2017; 2017:1042606. [PMID: 28512643 PMCID: PMC5415861 DOI: 10.1155/2017/1042606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/01/2017] [Indexed: 12/31/2022] Open
Abstract
Disaccharidases (DS) are brush border enzymes embedded in the microvillous membrane of small intestinal enterocytes. In untreated coeliac disease (CD), a general decrease of DS activities is seen. This manuscript reviews different aspects of DS activities in CD: their utility in the diagnosis and their application to in vitro toxicity testing. The latter has never been established in CD research. However, with the recent advances in small intestinal organoid techniques, DS might be employed as a biomarker for in vitro studies. This includes establishment of self-renewing epithelial cells raised from tissue, which express differentiation markers, including the brush border enzymes. Determining duodenal DS activities may provide additional information during the diagnostic workup of CD: (i) quantify the severity of the observed histological lesions, (ii) provide predictive values for the grade of mucosal villous atrophy, and (iii) aid diagnosing CD where minor histological changes are seen. DS can also provide additional information to assess the response to a gluten-free diet as marked increase of their activities occurs four weeks after commencing it. Various endogenous and exogenous factors affecting DS might also be relevant when considering investigating the role of DS in other conditions including noncoeliac gluten sensitivity and DS deficiencies.
Collapse
|
30
|
von Martels JZH, Sadaghian Sadabad M, Bourgonje AR, Blokzijl T, Dijkstra G, Faber KN, Harmsen HJM. The role of gut microbiota in health and disease: In vitro modeling of host-microbe interactions at the aerobe-anaerobe interphase of the human gut. Anaerobe 2017; 44:3-12. [PMID: 28062270 DOI: 10.1016/j.anaerobe.2017.01.001] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/16/2016] [Accepted: 01/02/2017] [Indexed: 02/07/2023]
Abstract
The microbiota of the gut has many crucial functions in human health. Dysbiosis of the microbiota has been correlated to a large and still increasing number of diseases. Recent studies have mostly focused on analyzing the associations between disease and an aberrant microbiota composition. Functional studies using (in vitro) gut models are required to investigate the precise interactions that occur between specific bacteria (or bacterial mixtures) and gut epithelial cells. As most gut bacteria are obligate or facultative anaerobes, studying their effect on oxygen-requiring human gut epithelial cells is technically challenging. Still, several (anaerobic) bacterial-epithelial co-culture systems have recently been developed that mimic host-microbe interactions occurring in the human gut, including 1) the Transwell "apical anaerobic model of the intestinal epithelial barrier", 2) the Host-Microbiota Interaction (HMI) module, 3) the "Human oxygen-Bacteria anaerobic" (HoxBan) system, 4) the human gut-on-a-chip and 5) the HuMiX model. This review discusses the role of gut microbiota in health and disease and gives an overview of the characteristics and applications of these novel host-microbe co-culture systems.
Collapse
Affiliation(s)
- Julius Z H von Martels
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Mehdi Sadaghian Sadabad
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tjasso Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Russo I, Zeppa P, Iovino P, Del Giorno C, Zingone F, Bucci C, Puzziello A, Ciacci C. The culture of gut explants: A model to study the mucosal response. J Immunol Methods 2016; 438:1-10. [PMID: 27475701 DOI: 10.1016/j.jim.2016.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/16/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023]
Abstract
Various experimental model designs have been used to analyze the inflammatory pathways in human gastrointestinal illnesses. Traditionally, analytical techniques and animal models are popular experimental tools to study the inflammation process of intestinal diseases. However, the comparison of results between animal and human models is difficult for the inconsistency of outcomes. Although there are different animal models for studying the intestinal diseases, none of them fully represents the physiological and environmental conditions typical of the human species. Also, there is currently a concerted effort to decrease the experimental use of animals. On the converse, experimental protocols using the culture of gut mucosa had become popular with the advent of endoscopy which allows explanting multiple fragments from the intestine. The peculiar characteristic of this model is the ability to preserve in vitro the features that we found in vivo, thus also the response to various stimuli that differs from person to person. The aim of the present paper is to provide a review of some of the possible uses of the organ intestinal mucosa culture.
Collapse
Affiliation(s)
- Ilaria Russo
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Pio Zeppa
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Paola Iovino
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Chiara Del Giorno
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Fabiana Zingone
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Cristina Bucci
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Alessandro Puzziello
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy
| | - Carolina Ciacci
- Department of Medicine and Surgery, University Hospital San Giovanni di Dio e Ruggi d'Aragona, University of Salerno, Italy.
| |
Collapse
|
32
|
Abstract
Studies on bacterial enterotoxin-epithelium interactions require model systems capable of mimicking the events occurring at the molecular and cellular levels during intoxication. In this chapter, we describe organ culture as an often neglected alternative to whole-animal experiments or enterocyte-like cell lines. Like cell culture, organ culture is versatile and suitable for studying rapidly occurring events, such as enterotoxin binding and uptake. In addition, it is advantageous in offering an epithelium with more authentic permeability/barrier properties than any cell line, as well as a subepithelial lamina propria, harboring the immune cells of the gut mucosa.
Collapse
|
33
|
Picariello G, Ferranti P, Addeo F. Use of brush border membrane vesicles to simulate the human intestinal digestion. Food Res Int 2016. [DOI: 10.1016/j.foodres.2015.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Costa MO, Harding JCS, Hill JE. Development and evaluation of a porcine in vitro colon organ culture technique. In Vitro Cell Dev Biol Anim 2016; 52:942-952. [PMID: 27338737 DOI: 10.1007/s11626-016-0060-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/01/2016] [Indexed: 12/17/2022]
Abstract
The intestinal mucosa comprises a complex assemblage of specialized tissues that interact in numerous ways. In vitro cell culture models are generally focused on recreating a specific characteristic of this organ and do not account for the many interactions between the different tissues. In vitro organ culture (IVOC) methods offer a way to overcome these limitations, but prolonging cell viability is essential. This study aimed to determine the feasibility and optimal conditions for in vitro culture of swine colonic mucosa for use as an enteric pathogen infection model. Explants (n = 168) from commercial pigs (n = 12), aged 5 to 10 wk, were used to assess the impact of various culture protocols on explant viability. Explants were cultured for up to 5 d and formalin fixed at 24-h intervals. Following establishment of the culture protocol, explants (n = 208) from 13 pigs were evaluated at Day 0 and 5 of culture. Assessment of viability was based on histological changes (tissue architecture evaluated by H&E, immunostaining of cell proliferation marker Ki-67) and expression of genes encoding IL-1α, IL-8, TNF-α, IFN-γ, and e-cadherin. After 5 d in culture, 20% of explants displayed over 80% of epithelial coverage, whereas 31% of explants had more than 50% of their surface covered by columnar epithelium, and 81% had crypts but with a decreased number of Ki-67-positive cells when compared to Day 0. Notably, large variability in explant quality was observed between donor pigs. Best possible explants were obtained from the distal colon of pigs, processed immediately after euthanasia, cultured at the liquid-tissue-gas interface in media supplemented with a mixture of antibiotics and antifungals and an oxygen-rich gas mix.
Collapse
Affiliation(s)
- Matheus O Costa
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - John C S Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Janet E Hill
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada.
| |
Collapse
|
35
|
Vadstrup K, Galsgaard ED, Gerwien J, Vester-Andersen MK, Pedersen JS, Rasmussen J, Neermark S, Kiszka-Kanowitz M, Jensen T, Bendtsen F. Validation and Optimization of an Ex Vivo Assay of Intestinal Mucosal Biopsies in Crohn's Disease: Reflects Inflammation and Drug Effects. PLoS One 2016; 11:e0155335. [PMID: 27171179 PMCID: PMC4865152 DOI: 10.1371/journal.pone.0155335] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 04/27/2016] [Indexed: 12/22/2022] Open
Abstract
Crohn’s disease (CD) is a chronic illness demanding better therapeutics. The marketed biologics only benefit some patients or elicit diminishing effect over time. To complement the known methods in drug development and to obtain patient specific drug responses, we optimized and validated a known human explant method to test drug candidates and pathophysiological conditions in CD intestinal biopsies. Mucosal biopsies from 27 CD patients and 6 healthy individuals were collected to validate an explant assay test where the polarized tissue was cultured on a novel metal mesh disk, slightly immersed in medium imitating an air-liquid interphase. After culture in high oxygen for 24 hours with or without biological treatment in the medium, biopsy integrity and penetration of antibodies was measured by immunohistochemistry (IHC). Nine cytokines were quantified in the conditioned medium as a read-out for degree of inflammation in individual biopsies and used to evaluate treatment efficacy. The biopsies were well-preserved, showing few structural changes. IHC revealed tissue penetration of antibodies demonstrating ability to test therapeutic antibodies. The cytokine release to the medium showed that the assay can distinguish between inflammation states and then validate the known effect of two treatment biologics confirmed by a detection panel of five specific cytokines. Our data also suggest that the assay would be able to indicate which patients are responders to anti-TNF-α therapeutics, and which are non-responders. This study demonstrates this version of an ex vivo culture as a valid and robust assay to assess inflammation in mucosal biopsies and test of the efficacy of novel drug candidates and current treatments on individual patients–potentially for a personalized medicine approach.
Collapse
Affiliation(s)
- Kasper Vadstrup
- Gastro Unit, Medical Division, Hvidovre University Hospital, Hvidovre, Denmark
- Faculty of Health Sciences, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
- Biopharmaceutical Research Unit, Novo Nordisk A/S, Maaloev, Denmark
- * E-mail:
| | | | - Jens Gerwien
- Biopharmaceutical Research Unit, Novo Nordisk A/S, Maaloev, Denmark
| | | | | | - Julie Rasmussen
- Gastro Unit, Medical Division, Hvidovre University Hospital, Hvidovre, Denmark
| | - Søren Neermark
- Gastro Unit, Medical Division, Hvidovre University Hospital, Hvidovre, Denmark
| | | | - Teis Jensen
- Biopharmaceutical Research Unit, Novo Nordisk A/S, Maaloev, Denmark
| | - Flemming Bendtsen
- Gastro Unit, Medical Division, Hvidovre University Hospital, Hvidovre, Denmark
- Faculty of Health Sciences, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
36
|
Dedhia PH, Bertaux-Skeirik N, Zavros Y, Spence JR. Organoid Models of Human Gastrointestinal Development and Disease. Gastroenterology 2016; 150:1098-1112. [PMID: 26774180 PMCID: PMC4842135 DOI: 10.1053/j.gastro.2015.12.042] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 12/23/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022]
Abstract
We have greatly advanced our ability to grow a diverse range of tissue-derived and pluripotent stem cell-derived gastrointestinal (GI) tissues in vitro. These systems, broadly referred to as organoids, have allowed the field to move away from the often nonphysiological, transformed cell lines that have been used for decades in GI research. Organoids are derived from primary tissues and have the capacity for long-term growth. They contain varying levels of cellular complexity and physiological similarity to native organ systems. We review the latest discoveries from studies of tissue-derived and pluripotent stem cell-derived intestinal, gastric, esophageal, liver, and pancreatic organoids. These studies have provided important insights into GI development, tissue homeostasis, and disease and might be used to develop personalized medicines.
Collapse
Affiliation(s)
- Priya H. Dedhia
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA,Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nina Bertaux-Skeirik
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yana Zavros
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio.
| | - Jason R. Spence
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA,Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI 48109, USA,Authors for Correspondence: Jason R. Spence – , Twitter: @TheSpenceLab, Yana Zavros –
| |
Collapse
|
37
|
Le VH, Hernando LR, Lee C, Choi H, Jin Z, Nguyen KT, Go G, Ko SY, Park JO, Park S. Shape memory alloy-based biopsy device for active locomotive intestinal capsule endoscope. Proc Inst Mech Eng H 2016; 229:255-63. [PMID: 25834001 DOI: 10.1177/0954411915576946] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recently, capsule endoscopes have been used for diagnosis in digestive organs. However, because a capsule endoscope does not have a locomotive function, its use has been limited to small tubular digestive organs, such as small intestine and esophagus. To address this problem, researchers have begun studying an active locomotive intestine capsule endoscope as a medical instrument for the whole gastrointestinal tract. We have developed a capsule endoscope with a small permanent magnet that is actuated by an electromagnetic actuation system, allowing active and flexible movement in the patient's gut environment. In addition, researchers have noted the need for a biopsy function in capsule endoscope for the definitive diagnosis of digestive diseases. Therefore, this paper proposes a novel robotic biopsy device for active locomotive intestine capsule endoscope. The proposed biopsy device has a sharp blade connected with a shape memory alloy actuator. The biopsy device measuring 12 mm in diameter and 3 mm in length was integrated into our capsule endoscope prototype, where the device's sharp blade was activated and exposed by the shape memory alloy actuator. Then the electromagnetic actuation system generated a specific motion of the capsule endoscope to extract the tissue sample from the intestines. The final biopsy sample tissue had a volume of about 6 mm(3), which is a sufficient amount for a histological analysis. Consequently, we proposed the working principle of the biopsy device and conducted an in-vitro biopsy test to verify the feasibility of the biopsy device integrated into the capsule endoscope prototype using the electro-magnetic actuation system.
Collapse
Affiliation(s)
- Viet Ha Le
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | | | - Cheong Lee
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Hyunchul Choi
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Zhen Jin
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Kim Tien Nguyen
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Gwangjun Go
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Seong-Young Ko
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Jong-Oh Park
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| | - Sukho Park
- School of Mechanical Engineering, Chonnam National University, Gwangju, Korea
| |
Collapse
|
38
|
Danielsen EM, Christiansen N, Danielsen EM. Pasteurella multocida toxin: Targeting mast cell secretory granules during kiss-and-run secretion. Tissue Cell 2015; 48:1-9. [PMID: 26763205 DOI: 10.1016/j.tice.2015.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/23/2015] [Accepted: 12/23/2015] [Indexed: 12/19/2022]
Abstract
Pasteurella multocida toxin (PMT), a virulence factor of the pathogenic Gram-negative bacterium P. multocida, is a 146 kDa protein belonging to the A-B class of toxins. Once inside a target cell, the A domain deamidates the α-subunit of heterotrimeric G-proteins, thereby activating downstream signaling cascades. However, little is known about how PMT selects and enters its cellular targets. We therefore studied PMT binding and uptake in porcine cultured intestinal mucosal explants to identify susceptible cells in the epithelium and underlying lamina propria. In comparison with Vibrio cholera B-subunit, a well-known enterotoxin taken up by receptor-mediated endocytosis, PMT binding to the epithelial brush border was scarce, and no uptake into enterocytes was detected by 2h, implying that none of the glycolipids in the brush border are a functional receptor for PMT. However, in the lamina propria, PMT distinctly accumulated in the secretory granules of mast cells. This also occurred at 4 °C, ruling out endocytosis, but suggestive of uptake via pores that connect the granules to the cell surface. Mast cell granules are known to secrete their contents by a "kiss-and-run" mechanism, and we propose that PMT may exploit this secretory mechanism to gain entry into this particular cell type.
Collapse
Affiliation(s)
- Elisabeth M Danielsen
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Nina Christiansen
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - E Michael Danielsen
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
39
|
Michael Danielsen E, Hansen GH. Small molecule pinocytosis and clathrin-dependent endocytosis at the intestinal brush border: Two separate pathways into the enterocyte. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:233-43. [PMID: 26615917 DOI: 10.1016/j.bbamem.2015.11.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/20/2015] [Accepted: 11/21/2015] [Indexed: 12/30/2022]
Abstract
Pinocytosis at the small intestinal brush border was studied in postweaned porcine cultured mucosal explants, using the fluorescent polar probes Alexa hydrazide (AH, MW 570), Texas red dextran (TRD, MW ~ 3000), and Cascade blue dextran (CBD, MW ~ 10,000). Within 1 h, AH appeared in a string of subapical punctae in enterocytes, indicative of an ongoing constitutive pinocytosis. By comparison, TRD was taken up less efficiently into the same compartment, and no intracellular labeling of CBD was detectable, indicating that only small molecules are pinocytosed from the postweaned gut lumen. AH remained in the terminal web region in EEA-1-positive endosomes (“TWEEs”) for at least 2 h, implying that the pinocytic uptake does not proceed towards a transcytic pathway. Like AH, cholera toxin B subunit (CTB) was readily internalized, but the two probes appeared in completely non-overlapping subapical compartments, indicating the existence of two different uptake mechanisms operating simultaneously at the brush border. CTB is internalized by clathrin-dependent receptor mediated endocytosis, but surprisingly the toxin also caused a rapid disappearance from the apical cell surface of two major brush border enzymes, alkaline phosphatase and aminopeptidase N, demonstrating the disruptive effect of this pathway. By immunofluorescence, caveolin-1 was hardly detectable in enterocytes, arguing against a caveolae-mediated uptake of AH, whereas the pinocytosis/phagocytosis inhibitors dimethyl amiloride and cytochalasin D both arrested AH uptake. We propose that the constitutive pinocytic mechanism visualized by AH contributes to maintenance of membrane homeostasis and to enrich the contents of lipid raft constituents at the brush border.
Collapse
Affiliation(s)
- E Michael Danielsen
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark.
| | - Gert H Hansen
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health Sciences, University of Copenhagen, Denmark
| |
Collapse
|
40
|
Goodwin TJ, McCarthy M, Cohrs RJ, Kaufer BB. 3D tissue-like assemblies: A novel approach to investigate virus-cell interactions. Methods 2015; 90:76-84. [PMID: 25986169 PMCID: PMC5489059 DOI: 10.1016/j.ymeth.2015.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 01/09/2023] Open
Abstract
Virus-host cell interactions are most commonly analyzed in cells maintained in vitro as two-dimensional tissue cultures. However, these in vitro conditions vary quite drastically from the tissues that are commonly infected in vivo. Over the years, a number of systems have been developed that allow the establishment of three-dimensional (3D) tissue structures that have properties similar to their in vivo 3D counterparts. These 3D systems have numerous applications including drug testing, maintenance of large tissue explants, monitoring migration of human lymphocytes in tissues, analysis of human organ tissue development and investigation of virus-host interactions including viral latency. Here, we describe the establishment of tissue-like assemblies for human lung and neuronal tissue that we infected with a variety of viruses including the respiratory pathogens human parainfluenza virus type 3 (PIV3), respiratory syncytial virus (RSV) and SARS corona virus (SARS-CoV) as well as the human neurotropic herpesvirus, varicella-zoster virus (VZV).
Collapse
Affiliation(s)
- Thomas J Goodwin
- Disease Modelling/Tissue Analogues Laboratory, NASA Johnson Space Center, Houston, TX, USA.
| | - Maureen McCarthy
- Disease Modelling/Tissue Analogues Laboratory, NASA Johnson Space Center, Houston, TX, USA
| | - Randall J Cohrs
- Departments of Neurology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| | | |
Collapse
|
41
|
Cooper SEJ, Wilson S, Feighery CF. Twenty-Four Hour Ex Vivo Culture of Celiac Duodenal Biopsies. Methods Mol Biol 2015; 1326:47-51. [PMID: 26498611 DOI: 10.1007/978-1-4939-2839-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Organ culture is a valuable technique in celiac disease research. It provides the opportunity to examine interactions between different cell types during the disease process without the need for invasive in vivo studies. Biopsies are maintained in an oxygen-rich environment, in contact with, but not submerged in, culture medium. A very straightforward and successful method of organ culture is described here.
Collapse
Affiliation(s)
- Sarah E J Cooper
- Immunology Department, Trinity College Dublin, College Green, Dublin 2, Ireland.
| | - Sharon Wilson
- Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Conleth F Feighery
- Immunology Department, Trinity College Dublin, College Green, Dublin 2, Ireland. .,Immunology Department, St James Hospital, Dublin, Ireland.
| |
Collapse
|
42
|
Shimo S, Saitoh S, Saitoh Y, Ohno N, Ohno S. Morphological and immunohistochemical analyses of soluble proteins in mucous membranes of living mouse intestines by cryotechniques. Microscopy (Oxf) 2015; 64:189-203. [PMID: 25770106 DOI: 10.1093/jmicro/dfv012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/15/2015] [Indexed: 01/02/2023] Open
Abstract
We have performed immunohistochemical or ultrastructural analyses of living mouse small intestines using Epon blocks prepared by 'in vivo cryotechnique' (IVCT). By electron microscopy, intracellular ultrastructures of epithelial cells were well preserved in tissue areas 5-10 μm away from cryogen-contact surface tissues. Their microvilli contained dynamically waving actin filaments, and highly electron-dense organelles, such as mitochondria, were seen under the widely organized terminal web. By quick-freezing of fresh resected tissues (FT-QF), many erythrocytes were congested within blood vessels due to loss of blood pressure. By immersion-fixation (IM-DH) and perfusion-fixation (PF-DH), small vacuoles were often seen in the cytoplasm of epithelial cells, and their intercellular spaces were also dilated. Moreover, actin filament bundles were irregular in cross sections of microvilli, compared with those with IVCT. Epon-embedded thick sections were treated with sodium ethoxide, followed by antigen retrieval and immunostained for immunoglobulin A (IgA), Ig kappa light chain (Igκ), J-chain and albumin. By cryotechniques, IgA immunoreactivity was detected as tiny dot-like patterns in cytoplasm of some epithelial cells. Both J-chain and Igκ immunoreactivities were detected in the same local areas as those of IgA. By FT-QF, however, the IgA immunoreactivity was more weakly detected, compared with that with IVCT. In thick sections prepared by IM-DH and PF-DH, it was rarely observed in both plasma and epithelial cells. Another albumin was diffusely immunolocalized in extracellular matrices of mucous membranes and also within blood vessels. Thus, IVCT was useful for preservation of soluble proteins and ultrastructural analyses of dynamically changing epithelial cells of living mouse small intestines.
Collapse
Affiliation(s)
- Satoshi Shimo
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan Present address: Department of Occupational Therapy, Health Science University, 7187 Kodachi, Fujikawaguchiko, Yamanashi 401-0380, Japan
| | - Sei Saitoh
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Yurika Saitoh
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Nobuhiko Ohno
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Shinichi Ohno
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 409-3898, Japan
| |
Collapse
|
43
|
Enterohemorrhagic Escherichia coli colonization of human colonic epithelium in vitro and ex vivo. Infect Immun 2014; 83:942-9. [PMID: 25534942 PMCID: PMC4333473 DOI: 10.1128/iai.02928-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is an important foodborne pathogen causing gastroenteritis and more severe complications, such as hemorrhagic colitis and hemolytic uremic syndrome. Pathology is most pronounced in the colon, but to date there is no direct clinical evidence showing EHEC binding to the colonic epithelium in patients. In this study, we investigated EHEC adherence to the human colon by using in vitro organ culture (IVOC) of colonic biopsy samples and polarized T84 colon carcinoma cells. We show for the first time that EHEC colonizes human colonic biopsy samples by forming typical attaching and effacing (A/E) lesions which are dependent on EHEC type III secretion (T3S) and binding of the outer membrane protein intimin to the translocated intimin receptor (Tir). A/E lesion formation was dependent on oxygen levels and suppressed under oxygen-rich culture conditions routinely used for IVOC. In contrast, EHEC adherence to polarized T84 cells occurred independently of T3S and intimin and did not involve Tir translocation into the host cell membrane. Colonization of neither biopsy samples nor T84 cells was significantly affected by expression of Shiga toxins. Our study suggests that EHEC colonizes and forms stable A/E lesions on the human colon, which are likely to contribute to intestinal pathology during infection. Furthermore, care needs to be taken when using cell culture models, as they might not reflect the in vivo situation.
Collapse
|
44
|
Matsuoka Y, Tsujimoto Y. Role of RIP1 in physiological enterocyte turnover in mouse small intestine via nonapoptotic death. Genes Cells 2014; 20:11-28. [PMID: 25348793 DOI: 10.1111/gtc.12193] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 09/21/2014] [Indexed: 11/29/2022]
Abstract
Enterocyte shedding in the small intestine is often referred as an example of programmed cell death. However, little is known about the underlying mechanisms, although both apoptotic and nonapoptotic cell death have been suggested to play an important role. Here, we show by electron microscope that the majority of cells dying in the mouse small intestine do not display apoptotic characteristics. Chemical biological approach in vivo and in an organ culture showed that necrostatin-1 (Nec-1), an inhibitor of receptor-interacting protein 1 (RIP1, also called RIPK1), inhibited the shedding/nonapoptotic death of enterocyte, resulting in suppression of physiological enterocyte turnover. Moreover, RIP1 knockdown in vivo and RIP1 haploinsufficiency significantly suppressed physiological enterocyte turnover. Unlike Nec-1-sensitive (RIP1-dependent) cell death, so called necroptosis, which is also dependent on RIP3, physiological enterocyte turnover in RIP3-deficient mice was executed normally and still inhibited by Nec-1. As inhibition of the shedding/nonapoptotic death of enterocyte by Nec-1 resulted in suppression of crypt cell proliferation, the shedding process plays a dominant role over cell proliferation in maintaining homeostasis of enterocyte turnover. These results indicate that RIP1 plays a major role in physiological enterocyte turnover through a RIP3-independent nonapoptotic death mechanism in the mouse small intestine.
Collapse
Affiliation(s)
- Yosuke Matsuoka
- Department of Medical Genetics, Osaka University Medical School, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | |
Collapse
|
45
|
Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Biol 2014; 15:647-64. [PMID: 25237826 PMCID: PMC4352326 DOI: 10.1038/nrm3873] [Citation(s) in RCA: 525] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mammalian organs are challenging to study as they are fairly inaccessible to experimental manipulation and optical observation. Recent advances in three-dimensional (3D) culture techniques, coupled with the ability to independently manipulate genetic and microenvironmental factors, have enabled the real-time study of mammalian tissues. These systems have been used to visualize the cellular basis of epithelial morphogenesis, to test the roles of specific genes in regulating cell behaviours within epithelial tissues and to elucidate the contribution of microenvironmental factors to normal and disease processes. Collectively, these novel models can be used to answer fundamental biological questions and generate replacement human tissues, and they enable testing of novel therapeutic approaches, often using patient-derived cells.
Collapse
Affiliation(s)
- Eliah R Shamir
- Departments of Cell Biology and Oncology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| |
Collapse
|
46
|
Chernets N, Zhang J, Steinbeck MJ, Kurpad DS, Koyama E, Friedman G, Freeman TA. Nonthermal atmospheric pressure plasma enhances mouse limb bud survival, growth, and elongation. Tissue Eng Part A 2014; 21:300-9. [PMID: 25102046 DOI: 10.1089/ten.tea.2014.0039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The enhanced differentiation of mesenchymal cells into chondrocytes or osteoblasts is of paramount importance in tissue engineering and regenerative therapies. A newly emerging body of evidence demonstrates that appendage regeneration is dependent on reactive oxygen species (ROS) production and signaling. Thus, we hypothesized that mesenchymal cell stimulation by nonthermal (NT)-plasma, which produces and induces ROS, would (1) promote skeletal cell differentiation and (2) limb autopod development. Stimulation with a single treatment of NT-plasma enhanced survival, growth, and elongation of mouse limb autopods in an in vitro organ culture system. Noticeable changes included enhanced development of digit length and definition of digit separation. These changes were coordinated with enhanced Wnt signaling in the distal apical epidermal ridge (AER) and presumptive joint regions. Autopod development continued to advance for approximately 144 h in culture, seemingly overcoming the negative culture environment usually observed in this in vitro system. Real-time quantitative polymerase chain reaction analysis confirmed the up-regulation of chondrogenic transcripts. Mechanistically, NT-plasma increased the number of ROS positive cells in the dorsal epithelium, mesenchyme, and the distal tip of each phalange behind the AER, determined using dihydrorhodamine. The importance of ROS production/signaling during development was further demonstrated by the stunting of digital outgrowth when anti-oxidants were applied. Results of this study show NT-plasma initiated and amplified ROS intracellular signaling to enhance development of the autopod. Parallels between development and regeneration suggest that the potential use of NT-plasma could extend to both tissue engineering and clinical applications to enhance fracture healing, trauma repair, and bone fusion.
Collapse
Affiliation(s)
- Natalie Chernets
- 1 Electrical and Computer Engineering Department, Drexel University , Philadelphia, Pennsylvania
| | | | | | | | | | | | | |
Collapse
|
47
|
Mapes B, Chase M, Hong E, Ludvik A, Ceryes K, Huang Y, Kupfer SS. Ex vivo culture of primary human colonic tissue for studying transcriptional responses to 1α,25(OH)2 and 25(OH) vitamin D. Physiol Genomics 2014; 46:302-8. [PMID: 24550213 DOI: 10.1152/physiolgenomics.00194.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
1α,25-Dihydroxyvitamin D3 [1α,25(OH)2D3] is a steroid hormone derived from circulating 25(OH) vitamin D [25(OH)D] with chemopreventive effects in colorectal cancer. 1α,25(OH)2D3 acts through transcriptional mechanisms; however, our understanding of vitamin D transcriptional responses in the colon is derived from studies in transformed cancer cell lines which may not represent responses in normal healthy tissue. Here, we describe the optimization of an ex vivo culture model using primary colonic biopsy samples for studying short-term transcriptional response induced by 1α,25(OH)2D3 and 25(OH)D treatment. Colon biopsy samples from healthy subjects were maintained in primary culture and treated in parallel with 100 nM 1α,25(OH)2D3 or 62.5 nM 25(OH)D and vehicle control (ethanol). Viability was assessed using histology and enzymatic assays. Genome-wide transcriptional responses to 1α,25(OH)2D3 were assessed and expression of 25(OH)D targets CYP27B1 and CYP24A1 were measured by real time PCR. We show that ex vivo culture of colonic tissue remains viable for up to 8 h. The largest number of differentially expressed genes in response to 1α,25(OH)2D3 was noted after 6 h (n = 120). As proof of concept, the top upregulated gene was CYP24A1, a well-established vitamin D-responsive gene. With 25(OH)D treatment, mRNA expression of CYP27B1 was significantly increased after 1 h, while expression of CYP24A1 was greatest at 8 h. Ex vivo culture can be used to assess short-term transcriptional responses to 1α,25(OH)2D3 and 25(OH)D in primary tissue from human colon. Future studies will address interindividual differences in transcriptional responses.
Collapse
Affiliation(s)
- Brandon Mapes
- Section of Gastroenterology, Department of Medicine, University of Chicago Medical Center, Chicago, Illinois
| | | | | | | | | | | | | |
Collapse
|
48
|
Arakelyan A, Fitzgerald W, Grivel JC, Vanpouille C, Margolis L. Histocultures (tissue explants) in human retrovirology. Methods Mol Biol 2014; 1087:233-48. [PMID: 24158827 DOI: 10.1007/978-1-62703-670-2_19] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Viral pathogenesis is studied predominantly in cultures of primary isolated cells or cell lines. Many retroviruses efficiently replicate only in activated cells. Therefore, in order to become efficient viral producers cells should be artificially activated, a procedure which significantly changes cell physiology. However, for many viral diseases, like HIV-1 and other retroviruses' diseases, critical pathogenic events occur in tissues. Therefore, cell isolation from their native microenvironment prevents single-cell cultures from faithfully reflecting important aspects of cell-cell and cell-pathogen interactions that occur in the context of complex tissue cytoarchitecture. Tissue explants (histocultures) that retain tissue cytoarchitecture and many aspects of cell-cell interactions more faithfully represent in vivo tissue features. Human histocultures constitute an adequate model for studying viral pathogenesis under controlled laboratory conditions. Protocols for various human histocultures as applied to study retroviral pathogenesis, in particular of HIV-1, have been refined by our laboratory and are described in the present publication. Histocultures of human tonsils and lymph nodes, as well as of recto-sigmoid and cervicovaginal tissues can be used to study viral transmission, pathogenesis and as a preclinical platform for antivirals evaluation.
Collapse
Affiliation(s)
- Anush Arakelyan
- Section of Intercellular Interactions, Program on Physical Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
49
|
Tsilingiri K, Sonzogni A, Caprioli F, Rescigno M. A novel method for the culture and polarized stimulation of human intestinal mucosa explants. J Vis Exp 2013:e4368. [PMID: 23666550 DOI: 10.3791/4368] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Few models currently exist to realistically simulate the complex human intestine's micro-environment, where a variety of interactions take place. Proper homeostasis directly depends on these interactions, as they shape an entire immunological response inducing tolerance against food antigens while at the same time mounting effective immune responses against pathogenic microbes accidentally ingested with food. Intestinal homeostasis is preserved also through various complex interactions between the microbiota (including food-associated beneficial bacterial strains) and the host, that regulate the attachment/degradation of mucus, the production of antimicrobial peptides by the epithelial barrier, and the "education" of epithelial cells' that controls the tolerogenic or immunogenic phenotype of unique, gut-resident lymphoid cells' populations. These interactions have been so far very difficult to reproduce with in vitro assays using either cultured cell lines or peripheral blood mononuclear cells. In addition, mouse models differ substantially in components of the intestinal mucosa (mucus layer organization, commensal bacteria community) with respect to the human gut. Thus, studies of a variety of treatments to be brought in the clinics for important stress-related or pathological conditions such as irritable bowel syndrome, inflammatory bowel disease or colorectal cancer have been difficult to carry out. To address these issues, we developed a novel system that enables us to stimulate explants of human intestinal mucosa that retain their in situ conditioning by the host microbiota and immune response, in a polarized fashion. Polarized apical stimulation is of great importance for the outcome of the elicited immune response. It has been repeatedly shown that the same stimuli can produce completely different responses when they bypass the apical face of the intestinal epithelium, stimulating epithelial cells basolaterally or coming into direct contact with lamina propria components, switching the phenotype from tolerogenic to immunogenic and causing unnecessary and excessive inflammation in the area. We achieved polarized stimulation by gluing a cave cylinder which delimited the area of stimulation on the apical face of the mucosa as will be described in the protocol. We used this model to examine, among others, differential effects of three different Lactobacilli strains. We show that this model system is very powerful to assess the immunomodulatory properties of probiotics in healthy and disease conditions.
Collapse
|
50
|
Fang SB, Schüller S, Phillips AD. Human Intestinal In Vitro Organ Culture as a Model for Investigation of Bacteria–Host Interactions. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.jecm.2013.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|